Abstract

Simple SummaryLung cancer (LC) is the tumor with the highest global mortality rate. Novel personalized therapies are currently being tested (e.g., targeted inhibitors, the immune-checkpoint inhibitors), but they cannot yet prevent the very frequent relapse and generalized metastases observed in a large population of LC patients. Currently, there is an urgent need for novel reliable biomarkers for the prognosis and diagnosis of LC. Through the systematic analysis of multiple deposited expression datasets, this report aims to explore the role of the Yin-Yang 1 (YY1) transcription factor and its target the Raf Kinase Inhibitory Protein (RKIP) in LC. The computational analysis suggested the predictive diagnostic and prognostic roles for both YY1 and RKIP stimulating further studies for proving their implication as novel biomarkers, as well as therapeutically druggable targets in LC.Lung cancer (LC) represents a global threat, being the tumor with the highest mortality rate. Despite the introduction of novel therapies (e.g., targeted inhibitors, immune-checkpoint inhibitors), relapses are still very frequent. Accordingly, there is an urgent need for reliable predictive biomarkers and therapeutically druggable targets. Yin-Yang 1 (YY1) is a transcription factor that may work either as an oncogene or a tumor suppressor, depending on the genotype and the phenotype of the tumor. The Raf Kinase Inhibitory Protein (RKIP), is a tumor suppressor and immune enhancer often found downregulated in the majority of the examined cancers. In the present report, the role of both YY1 and RKIP in LC is thoroughly explored through the analysis of several deposited RNA and protein expression datasets. The computational analyses revealed that YY1 negatively regulates RKIP expression in LC, as corroborated by the deposited YY1-ChIP-Seq experiments and validated by their robust negative correlation. Additionally, YY1 expression is significantly higher in LC samples compared to normal matching ones, whereas RKIP expression is lower in LC and high in normal matching tissues. These observed differences, unlike many current biomarkers, bear a diagnostic significance, as proven by the ROC analyses. Finally, the survival data support the notion that both YY1 and RKIP might represent strong prognostic biomarkers. Overall, the reported findings indicate that YY1 and RKIP expression levels may play a role in LC as potential biomarkers and therapeutic targets. However, further studies will be necessary to validate the in silico results.

Highlights

  • Lung cancer (LC) represents the second most widely diagnosed cancer, as well as the first cause of death due to a malignancy

  • The analysis showed that Yin-Yang 1 (YY1) may bind the promoter of Raf Kinase Inhibitory Protein (RKIP) at the level of seven different binding sequences, with a relative score included between 80.4% and 87.5% (Figure 1C)

  • Supplementary Table S1, the experiments demonstrated the existence of nine different binding clusters located between −15,000 bp and +5000 bp around the RKIP transcription starting site (TSS), for a total of 23 binding peaks, each one corresponding to the binding of YY1 TF to the DNA of the RKIP gene regulatory region

Read more

Summary

Introduction

Lung cancer (LC) represents the second most widely diagnosed cancer, as well as the first cause of death due to a malignancy. An estimate of 2.2 million new LC diagnoses and about 1.8 million deaths for LC were reported during the year 2020 [1]. The most diffused form of LC is non-small cell lung cancer (NSCLC), with a prevalence of about. The remaining 10% of LC cases are represented by small cell lung cancer (SCLC) [2]. The 5-year survival rate for individuals diagnosed with LC varies between 5% and 20%, depending on the tumor stage, as well as the geographical area, underlining the major role played by the individual genetic background [1]. Novel diagnostic approaches, including low-dose computed tomography (LDCT) for high-risk subjects, are forestalling the time of diagnosis [6–8]

Objectives
Methods
Results
Discussion
Conclusion
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call